Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Appl Microbiol Biotechnol ; 102(16): 7047-7059, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29931601

ABSTRACT

Interferon beta (IFNß) is transiently expressed in response to viral infections and widely used to treat relapsing-remitting multiple sclerosis (MS). We introduced mutations in the IFNß gene (in the 27th and 101st codons and in the Kozak sequence, and also deletion of 3' and 5' unstable, untranslated region, UTR) with the aim of increasing the expression of IFNß. Computational analyses of mutant and wild-type RNAs and proteins of IFNß by RNAfold, ASAView, HOPE and Ramachandran plot, and iStable web servers showed that the mutations could decrease RNA stability, protein solvent accessibility, and protein stability but could not change correct folding. Two recombinant IFNß101 and IFNß101+27 constructs were designed by site-directed mutagenesis. The wild-type IFNß gene also was used as a control. In vitro experiments by quantitative real-time PCR, dot blot, SDS-PAGE, and Western blot assays showed an increased expression level of recombinant IFNßs. 79.9-fold, 99.7-fold, and 190-fold elevations in the mRNA expression of IFNßw, IFNß101, and IFNß101+27 were seen, respectively, in comparison with the endogenous IFNß mRNA in untransfected HEK293T cells. The IFNß mRNA expression was increased 2.38-fold and 1.25-fold for 101+27 and 101 mutated forms, respectively, in comparison with the IFNß wild-type construct. An elevation in IFNß protein production was also clearly detected in the transfected HEK293T cell containing recombinant IFNß101 and IFNß101+27 constructs. Finally, these directed mutations in the IFNß gene successfully elevated protein and mRNA production but in silico analyses of mutant mRNAs showed decreased mRNA stability, unlike previous studies, in comparison with the wild-type mRNA.


Subject(s)
Interferon beta-1a/biosynthesis , Interferon beta-1a/genetics , Computer Simulation , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Protein Folding , Protein Stability , RNA Stability , Recombinant Proteins
2.
J Immunol ; 199(8): 2618-2623, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28904124

ABSTRACT

The transitional stage of B cell development is a formative stage in the spleen where autoreactive specificities are censored as B cells gain immune competence, but the intrinsic and extrinsic factors regulating survival of transitional stage 1 (T1) B cells are unknown. We report that B cell expression of IFN-ß is required for optimal survival and TLR7 responses of transitional B cells in the spleen and was overexpressed in T1 B cells from BXD2 lupus-prone mice. Single-cell gene expression analysis of B6 Ifnb+/+ versus B6 Ifnb-/- T1 B cells revealed heterogeneous expression of Ifnb in wild-type B cells and distinct gene expression patterns associated with endogenous IFN-ß. Single-cell analysis of BXD2 T1 B cells revealed that Ifnb is expressed in early T1 B cell development with subsequent upregulation of Tlr7 and Ifna1 Together, these data suggest that T1 B cell expression of IFN-ß plays a key role in regulating responsiveness to external factors.


Subject(s)
B-Lymphocytes/immunology , Interferon-beta/metabolism , Lupus Nephritis/immunology , Precursor Cells, B-Lymphoid/immunology , Spleen/immunology , Animals , B-Lymphocyte Subsets/immunology , Cell Differentiation , Cell Survival , Disease Susceptibility , Interferon beta-1a/genetics , Interferon beta-1a/metabolism , Interferon-alpha , Lymphocyte Activation , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred Strains , Single-Cell Analysis , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism
3.
Cell Rep ; 17(2): 425-435, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27705791

ABSTRACT

Functional genomic analysis of gene expression in mice allowed us to identify a quantitative trait locus (QTL) linked in trans to the expression of 190 gene transcripts and in cis to the expression of only two genes, one of which was Ypel5. Most of the trans-expression QTL genes were interferon-stimulated genes (ISGs), and their expression in mouse macrophage cell lines was stimulated in an IFNB1-dependent manner by Ypel5 silencing. In human HEK293T cells, YPEL5 silencing enhanced the induction of IFNB1 by pattern recognition receptors and phosphorylation of TBK1/IKBKE kinases, whereas co-immunoprecipitation experiments revealed that YPEL5 interacted physically with IKBKE. We thus found that the Ypel5 gene (contained in a locus linked to a network of ISGs in mice) is a negative regulator of IFNB1 production and innate immune responses that interacts functionally and physically with TBK1/IKBKE kinases.


Subject(s)
Cell Cycle Proteins/genetics , I-kappa B Kinase/genetics , Interferon beta-1a/genetics , Interferons/genetics , Protein Serine-Threonine Kinases/genetics , Animals , HEK293 Cells , Humans , Macrophages/metabolism , Mice , Protein Binding , Quantitative Trait Loci/genetics , Regulatory Elements, Transcriptional/genetics
4.
J Immunol ; 197(6): 2338-52, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27511736

ABSTRACT

Type I IFN (IFN-α/ß)-driven immune responses to acute viral infection are critical to counter replication and prevent dissemination. However, the mechanisms underlying host resistance to HSV type 1 (HSV-1) are incompletely understood. In this study, we show that mice with deficiencies in IFN-α/ß signaling or stimulator of IFN genes (STING) exhibit exacerbated neurovirulence and atypical lymphotropic dissemination of HSV-1 following ocular infection. Synergy between IFN-α/ß signaling and efficacy of early adaptive immune responses to HSV-1 were dissected using bone marrow chimeras and adoptive cell transfer approaches to profile clonal expansion, effector function, and recruitment of HSV-specific CD8(+) T cells. Lymphotropic viral dissemination was commensurate with abrogated CD8(+) T cell responses and pathological alterations of fibroblastic reticular cell networks in the draining lymph nodes. Our results show that resistance to HSV-1 in the trigeminal ganglia during acute infection is conferred in part by STING and IFN-α/ß signaling in both bone marrow-derived and -resident cells, which coalesce to support a robust HSV-1-specific CD8(+) T cell response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Interferon beta-1a/metabolism , Interferon-alpha/metabolism , Lymphadenitis/immunology , Lymphadenitis/virology , Membrane Proteins/metabolism , Adaptive Immunity , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/pathology , Eye/virology , Herpes Simplex/immunology , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/physiology , Interferon beta-1a/genetics , Interferon beta-1a/immunology , Interferon-alpha/genetics , Interferon-alpha/immunology , Lymphadenitis/physiopathology , Membrane Proteins/deficiency , Mice , Signal Transduction , Trigeminal Ganglion/immunology , Trigeminal Ganglion/physiopathology , Trigeminal Ganglion/virology , Virus Replication
5.
BMC Pharmacol Toxicol ; 16: 25, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26392348

ABSTRACT

BACKGROUND: The innate immune receptor RIG-I detects viral RNA within the cytosol of infected cells. Activation of RIG-I leads to the induction of antiviral cytokines, in particular type I interferon, the inhibition of a T(H)17 response as well as to the suppression of tumor growth. Therefore, RIG-I is a promising drug target for the treatment of cancer as well as multiple sclerosis. A specific ligand for RIG-I is currently in preclinical testing. The first-in-human trial will need to be carefully designed to avoid an overshooting cytokine response. Therefore, the ResI study was set up to analyze the human immune response to standard treatment with recombinant interferon-beta to establish biomarkers for safety and efficacy of the upcoming first-in-human trial investigating the RIG-I ligand. METHODS/DESIGN: ResI is a single center, prospective, open label, non-randomized phase I clinical trial. Three different cohorts (20 healthy volunteers, 20 patients with RRMS and ongoing interferon-beta treatment and 10 patients starting on interferon-beta) will receive standard interferon-beta-1a therapy for nine days. The study will be conducted according to the principles of the german medicinal products act, ICH-GCP, and the Declaration of Helsinki on the phase I unit of the Institute of Clinical Chemistry and Clinical Pharmacology and in the Department of Neurology, both University Hospital Bonn. Interferon-beta-induced cytokine levels, surface marker on immune cells, mRNA- and miRNA-expression as well as psychometric response will be investigated as target variables. DISCUSSION: The ResI study will assess biomarkers in response to interferon-ß treatment to guide the dose steps within the first-in-human trial with a newly developed RIG-I ligand. Thus, ResI is a biomarker study to enhance the safety of the clinical development of a first-in-class compound. The data can additionally be used for the development of other therapies based on type I interferon induction such as TLR ligands. Moreover, it will help to understand the interferon-beta induced immune response in a controlled in vivo setting in the human system. TRIAL REGISTRATION: clinicaltrials.gov ID NCT02364986.


Subject(s)
Interferon Type I/metabolism , Interferon beta-1a/therapeutic use , MicroRNAs/genetics , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/therapeutic use , Adolescent , Adult , Aged , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Gene Expression/drug effects , Humans , Interferon beta-1a/genetics , Interferon beta-1a/metabolism , Male , MicroRNAs/metabolism , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/metabolism , Outcome Assessment, Health Care , Prospective Studies , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Immunologic , Recombinant Proteins/therapeutic use , Time Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...